1
|
Thi EP, Ye X, Snead NM, Lee ACH, Micolochick Steuer HM, Ardzinski A, Graves IE, Espiritu C, Cuconati A, Abbott C, Jarosz A, Teng X, Paratala B, McClintock K, Harasym T, Rijnbrand R, Lam AM, Sofia MJ. Control of Hepatitis B Virus with Imdusiran, a Small Interfering RNA Therapeutic. ACS Infect Dis 2024; 10:3640-3649. [PMID: 39306863 DOI: 10.1021/acsinfecdis.4c00514] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Chronic hepatitis B is a global health concern with a high risk of end-stage liver disease. Current standard-of-care agents have low cure rates, and new therapies are needed. Small interfering RNAs (siRNAs) that target viral RNAs fulfill a gap not addressed by standard-of-care agents and may contribute to a functional cure. Here, we describe the preclinical characterization of imdusiran (AB-729), a novel, pan-genotypic siRNA therapeutic that effectively reduces HBsAg, viral antigens, and viral replication in chronic hepatitis B patients and is currently in Phase 2 clinical studies. In hepatitis B virus (HBV) cell-based systems, imdusiran possessed pan-genotypic nanomolar potency and retained activity against HBV target site polymorphisms. Imdusiran was active against nucleos(t)ide analogue- and capsid assembly modulator-resistant HBV isolates, and combination with standard-of-care agents was additive. In an HBV adeno-associated virus mouse model, HBsAg was reduced up to 3.7 log10 after a single imdusiran dose, with sustained suppression for 10 weeks. Imdusiran did not intrinsically stimulate cytokine release in healthy donor human whole blood, supportive of its mechanism of action as a direct acting RNA interference antiviral. Taken together, these data support imdusiran in combination treatment approaches toward chronic hepatitis B functional cure.
Collapse
Affiliation(s)
- Emily P Thi
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Xin Ye
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Nicholas M Snead
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Amy C H Lee
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | | | - Andrzej Ardzinski
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Ingrid E Graves
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Christine Espiritu
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Andrea Cuconati
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Cory Abbott
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Agnes Jarosz
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Xiaowei Teng
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Bhavna Paratala
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Kevin McClintock
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Troy Harasym
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Rene Rijnbrand
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Angela M Lam
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Michael J Sofia
- Arbutus Biopharma Inc. 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| |
Collapse
|
2
|
Liu D, Li T, Liu L, Che X, Li X, Liu C, Wu G. Adeno-associated virus therapies: Pioneering solutions for human genetic diseases. Cytokine Growth Factor Rev 2024:S1359-6101(24)00078-9. [PMID: 39322487 DOI: 10.1016/j.cytogfr.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
Adeno-associated virus (AAV) has emerged as a fundamental component in the gene therapy landscape, widely acknowledged for its effectiveness in therapeutic gene delivery. The success of AAV-based therapies, such as Luxturna and Zolgensma, underscores their potential as a leading vector in gene therapy. This article provides an in-depth review of the development and mechanisms of AAV vector-based therapies, offering a comprehensive analysis of the latest clinical trial outcomes in central nervous system (CNS) diseases, ocular conditions, and hemophilia, where AAV therapies have shown promising results. Additionally, we discusse the selection of administration methods and serotypes tailored to specific diseases. Our objective is to showcase the innovative applications and future potential of AAV-based gene therapy, laying the groundwork for continued clinical advancements.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaorui Li
- Department of oncology, Cancer Hospital of Dalian University of Technology, Shenyang 110042, China.
| | - Chang Liu
- Department of thoracic surgery, Shenyang Tenth People's Hospital, Shenyang 110042, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
3
|
Bailey JT, Cangialosi S, Moshkani S, Rexhouse C, Cimino JL, Robek MD. CD40 stimulation activates CD8+ T cells and controls HBV in CD4-depleted mice. JHEP Rep 2024; 6:101121. [PMID: 39282227 PMCID: PMC11399595 DOI: 10.1016/j.jhepr.2024.101121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/14/2024] [Indexed: 09/18/2024] Open
Abstract
Background & Aims HBV treatment is challenging due to the persistence of the covalently closed circular DNA replication pool, which remains unaffected by antiviral intervention. In this study, we determined whether targeting antigen-presenting cells via CD40 stimulation represents an appropriate therapeutic approach for achieving sustained HBV control in a mouse model of HBV replication. Methods Mice were transduced with an adeno-associated virus encoding the HBV genome (AAV-HBV) to initiate HBV replication and were administered agonistic CD40 antibody. CD4-depleting antibody was administered in addition to the CD40 antibody. Viral antigens in the blood were measured over time to determine HBV control. HBV-specific CD8+ T cells were quantified in the spleen and liver at the experimental endpoint. Results CD40 stimulation in CD4-depleted AAV-HBV mice resulted in the clearance of HBsAg and HBeAg, along with a reduction in liver HBV mRNA, contrasting with CD4-competent counterparts. CD8+ T cells were indispensable for CD40-mediated HBV control, determined by HBV persistence following their depletion. In CD4-replete mice, CD40 stimulation initially facilitated the expansion of HBV-specific CD8+ T cells, which subsequently could not control HBV. Finally, α-CD4/CD40 treatment reduced antigenemia and liver HBV mRNA levels in chronic AAV-HBV mice, with further enhancement through synergy with immunization by VSV-MHBs (vesicular stomatitis virus expressing middle HBsAg). Conclusions Our findings underscore the potential of CD40 stimulation as a targeted therapeutic strategy for achieving sustained HBV control and reveal a CD4+ T cell-dependent limitation on CD40-mediated antiviral efficacy. Impact and implications Immunotherapy has the potential to overcome immune dysfunction in chronic HBV infection. Using a mouse model of HBV replication, this study shows that CD40 stimulation can induce sustained HBV control, which is dependent on CD8+ T cells and further enhanced by co-immunization. Unexpectedly, CD40-mediated HBV reduction was improved by the depletion of CD4+ cells. These findings suggest potential strategies for reversing HBV persistence in infected individuals.
Collapse
Affiliation(s)
- Jacob T Bailey
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Sophia Cangialosi
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Safiehkhatoon Moshkani
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Catherine Rexhouse
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Jesse L Cimino
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Michael D Robek
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
4
|
Xu X, Liu J, Li X, Feng Q, Su Y. Integrated network pharmacology and metabolomics to study the potential mechanism of Jiawei Yinchenhao decoction in chronic hepatitis B. Heliyon 2024; 10:e36267. [PMID: 39224343 PMCID: PMC11367511 DOI: 10.1016/j.heliyon.2024.e36267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Chronic hepatitis B infection (CHB) is a major risk factor for the development of hepatocellular carcinoma (HCC) globally and continues to pose a significant global health challenge. Jiawei Yinchenhao decoction (JWYCH) is a modified version of Yinchenhao decoction (YCHD), which is widely used to treat liver diseases including icteric hepatitis, cholelithiasis, and hepatic ascites. However, the effectiveness and underlying mechanism of JWYCH on CHB are still unclear. This study aimed to investigate the impact of JWYCH on CHB and explore the underlying mechanism via network pharmacology and metabolomics. C57BL/6 mice were administered rAAV-HBV1.3 via hydrodynamic injection (HDI) to establish the CHB model. The infected mice were orally administered JWYCH for 4 weeks. HBsAg, HBeAg, HBV DNA, the serum liver function index, and histopathology were detected. In addition, network pharmacology was used to investigate potential targets, whereas untargeted metabolomics analysis was employed to explore the hepatic metabolic changes in JWYCH in CHB mice and identify relevant biomarkers and metabolic pathways. JWYCH was able to reduce HBeAg levels and improve liver pathological changes in mice with CHB. Additionally, metabolomics analysis indicated that JWYCH can influence 105 metabolites, including pipecolic acid, alpha-terpinene, adenosine, and L-phenylalanine, among others. Bile acid metabolism, arachidonic acid metabolism, and retinol metabolism are suggested to be potential targets of JWYCH in CHB. In conclusion, JWYCH demonstrated a hepatoprotective effect on a mouse model of CHB, suggesting a potential alternative therapeutic strategy for CHB. The effect of JWYCH is associated mainly with regulating the metabolism of bile acid, arachidonic acid, and retinol. These differentially abundant metabolites may serve as potential biomarkers and therapeutic targets for CHB.
Collapse
Affiliation(s)
- Xinyi Xu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jin Liu
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xue Li
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - QuanSheng Feng
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yue Su
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
5
|
Zhang T, Yang J, Gao H, Wu Y, Zhao X, Zhao H, Xie X, Yang L, Li Y, Wu Q. Progress of Infection and Replication Systems of Hepatitis B Virus. ACS Pharmacol Transl Sci 2024; 7:1711-1721. [PMID: 38898948 PMCID: PMC11184603 DOI: 10.1021/acsptsci.4c00147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
Despite the long-standing availability of effective prophylaxis, chronic hepatitis B virus (HBV) infection remains a formidable public health threat. Antiviral treatments can limit viral propagation, but prolonged therapy is necessary to control HBV replication. Robust in vitro models of HBV infection are indispensable prerequisites for elucidating viral pathogenesis, delineating virus-host interplay and developing novel therapeutic, preventative countermeasures. Buoyed by advances in molecular techniques and tissue culture systems, investigators have engineered numerous in vitro models of the HBV life cycle. However, all current platforms harbor limitations in the recapitulation of natural infection. In this article, we comprehensively review the HBV life cycle, provide an overview of existing in vitro HBV infection and replication systems, and succinctly present the benefits and caveats in each model with the primary objective of constructing refined experimental models that closely mimic native viral infection and offering robust support for the ambitious "elimination of hepatitis by 2030" initiative.
Collapse
Affiliation(s)
- Tiantian Zhang
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Juan Yang
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - He Gao
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Yuwei Wu
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Xinyu Zhao
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Hui Zhao
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Xinqiang Xie
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Lingshuang Yang
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Ying Li
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Qingping Wu
- National
Health Commission Science and Technology Innovation Platform for Nutrition
and Safety of Microbial Food, Guangdong Provincial Key Laboratory
of Microbial Safety and Health, State Key Laboratory of Applied Microbiology
Southern China, Institute of Microbiology,
Guangdong Academy of Sciences, Guangzhou 510070, China
| |
Collapse
|
6
|
Berke JM, Tan Y, Sauviller S, Wu DT, Zhang K, Conceição-Neto N, Blázquez Moreno A, Kong D, Kukolj G, Li C, Zhu R, Nájera I, Pauwels F. Class A capsid assembly modulator apoptotic elimination of hepatocytes with high HBV core antigen level in vivo is dependent on de novo core protein translation. J Virol 2024; 98:e0150223. [PMID: 38315015 PMCID: PMC10949496 DOI: 10.1128/jvi.01502-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/19/2023] [Indexed: 02/07/2024] Open
Abstract
Capsid assembly is critical in the hepatitis B virus (HBV) life cycle, mediated by the viral core protein. Capsid assembly is the target for new anti-viral therapeutics known as capsid assembly modulators (CAMs) of which the CAM-aberrant (CAM-A) class induces aberrant shaped core protein structures and leads to hepatocyte cell death. This study aimed to identify the mechanism of action of CAM-A modulators leading to HBV-infected hepatocyte elimination where CAM-A-mediated hepatitis B surface antigen (HBsAg) reduction was evaluated in a stable HBV replicating cell line and in AAV-HBV-transduced C57BL/6, C57BL/6 SCID, and HBV-infected chimeric mice with humanized livers. Results showed that in vivo treatment with CAM-A modulators induced pronounced reductions in hepatitis B e antigen (HBeAg) and HBsAg, associated with a transient alanine amino transferase (ALT) increase. Both HBsAg and HBeAg reductions and ALT increase were delayed in C57BL/6 SCID and chimeric mice, suggesting that adaptive immune responses may indirectly contribute. However, CD8+ T cell depletion in transduced wild-type mice did not impact antigen reduction, indicating that CD8+ T cell responses are not essential. Transient ALT elevation in AAV-HBV-transduced mice coincided with a transient increase in endoplasmic reticulum stress and apoptosis markers, followed by detection of a proliferation marker. Microarray data revealed antigen presentation pathway (major histocompatibility complex class I molecules) upregulation, overlapping with the apoptosis. Combination treatment with HBV-specific siRNA demonstrated that CAM-A-mediated HBsAg reduction is dependent on de novo core protein translation. To conclude, CAM-A treatment eradicates HBV-infected hepatocytes with high core protein levels through the induction of apoptosis, which can be a promising approach as part of a regimen to achieve functional cure. IMPORTANCE Treatment with hepatitis B virus (HBV) capsid assembly modulators that induce the formation of aberrant HBV core protein structures (CAM-A) leads to programmed cell death, apoptosis, of HBV-infected hepatocytes and subsequent reduction of HBV antigens, which differentiates CAM-A from other CAMs. The effect is dependent on the de novo synthesis and high levels of core protein.
Collapse
Affiliation(s)
- Jan Martin Berke
- Infectious Diseases Discovery, Infectious Diseases and Vaccines, Janssen Research and Development, Turnhoutseweg, Beerse, Belgium
| | - Ying Tan
- Infectious Diseases Discovery, Janssen Research and Development, Jinchuang Mansion, Pudong, Shanghai, China
| | - Sarah Sauviller
- Infectious Diseases Discovery, Infectious Diseases and Vaccines, Janssen Research and Development, Turnhoutseweg, Beerse, Belgium
| | - Dai-tze Wu
- Infectious Diseases Discovery, Janssen Research and Development, Jinchuang Mansion, Pudong, Shanghai, China
| | - Ke Zhang
- Infectious Diseases Discovery, Janssen Research and Development, Jinchuang Mansion, Pudong, Shanghai, China
| | - Nádia Conceição-Neto
- Infectious Diseases Discovery, Infectious Diseases and Vaccines, Janssen Research and Development, Turnhoutseweg, Beerse, Belgium
| | - Alfonso Blázquez Moreno
- Infectious Diseases Biomarkers, Infectious Diseases and Vaccines, Janssen Research and Development, Turnhoutseweg, Beerse, Belgium
| | - Desheng Kong
- Infectious Diseases Discovery, Janssen Research and Development, Jinchuang Mansion, Pudong, Shanghai, China
| | - George Kukolj
- Infectious Diseases Discovery, Janssen Research and Development, Brisbane, California, USA
| | - Chris Li
- Infectious Diseases Discovery, Janssen Research and Development, Brisbane, California, USA
| | - Ren Zhu
- Infectious Diseases Discovery, Janssen Research and Development, Jinchuang Mansion, Pudong, Shanghai, China
| | - Isabel Nájera
- Infectious Diseases Discovery, Janssen Research and Development, Brisbane, California, USA
| | - Frederik Pauwels
- Infectious Diseases Discovery, Infectious Diseases and Vaccines, Janssen Research and Development, Turnhoutseweg, Beerse, Belgium
| |
Collapse
|
7
|
Schiller L, Ko C, Kosinska AD, Grimm D, Protzer U. Production and Purification of Adeno-Associated Viral Vectors for the Development of Immune-Competent Mouse Models of Persistent Hepatitis B Virus Replication. Methods Mol Biol 2024; 2837:207-218. [PMID: 39044087 DOI: 10.1007/978-1-0716-4027-2_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Mice infected with a recombinant adeno-associated virus carrying a replication-competent hepatitis B virus genome (rAAV-HBV) via the intravenous route establish a persistent HBV replication in hepatocytes and develop immune tolerance. They serve as models to evaluate antiviral immunity and to assess potential therapeutic approaches for chronic HBV infection. Combining selected HBV variants and different mouse genotypes allows for addressing a broad spectrum of research questions. This chapter describes the basic principles of the rAAV-HBV mouse model, rAAV-HBV production and purification methods, and finally, the in vivo application.
Collapse
Affiliation(s)
- Lisa Schiller
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany
| | - Chunkyu Ko
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Anna D Kosinska
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner sites, Munich and Heidelberg, Germany
| | - Dirk Grimm
- German Center for Infection Research (DZIF), partner sites, Munich and Heidelberg, Germany
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany.
- German Center for Infection Research (DZIF), partner sites, Munich and Heidelberg, Germany.
| |
Collapse
|
8
|
Xu C, Fan J, Liu D, Tuerdi A, Chen J, Wei Y, Pan Y, Dang H, Wei X, Yousif AS, Yogaratnam J, Zhou Q, Lichenstein H, Xu T. Alpha-kinase 1 (ALPK1) agonist DF-006 demonstrates potent efficacy in mouse and primary human hepatocyte (PHH) models of hepatitis B. Hepatology 2023; 77:275-289. [PMID: 35699669 DOI: 10.1002/hep.32614] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIMS In the treatment of chronic hepatitis B (CHB) infection, stimulation of innate immunity may lead to hepatitis B virus (HBV) cure. Alpha-kinase 1 (ALPK1) is a pattern recognition receptor (PRR) that activates the NF-κB pathway and stimulates innate immunity. Here we characterized the preclinical anti-HBV efficacy of DF-006, an orally active agonist of ALPK1 currently in clinical development for CHB. APPROACH AND RESULTS In adeno-associated virus (AAV)-HBV mouse models and primary human hepatocytes (PHHs) infected with HBV, we evaluated the antiviral efficacy of DF-006. In the mouse models, DF-006 rapidly reduced serum HBV DNA, hepatitis B surface antigen, and hepatitis B e antigen levels using doses as low as 0.08 μg/kg, 1 μg/kg, and 5 μg/kg, respectively. DF-006 in combination with the HBV nucleoside reverse transcriptase inhibitor, entecavir, further reduced HBV DNA. Antiviral efficacy in mice was associated with an increase in immune cell infiltration and decrease of hepatitis B core antigen, encapsidated pregenomic RNA, and covalently closed circular DNA in liver. At subnanomolar concentrations, DF-006 also showed anti-HBV efficacy in PHH with significant reductions of HBV DNA. Following dosing with DF-006, there was upregulation of NF-κB-targeted genes that are involved in innate immunity. CONCLUSION DF-006 was efficacious in mouse and PHH models of HBV without any indications of overt toxicity. In mice, DF-006 localized primarily to the liver where it potently activated innate immunity. The transcriptional response in mouse liver provides insights into mechanisms that mediate anti-HBV efficacy by DF-006.
Collapse
Affiliation(s)
- Cong Xu
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
- Zhejiang Yao Yuan Biotechnology Ltd , Jiashan , Zhejiang , China
| | - Jieqing Fan
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
- Zhejiang Yao Yuan Biotechnology Ltd , Jiashan , Zhejiang , China
| | - Danyang Liu
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
- Zhejiang Yao Yuan Biotechnology Ltd , Jiashan , Zhejiang , China
| | - Aimaier Tuerdi
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
| | - Juanjuan Chen
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
| | - Yuning Wei
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
| | - Yanfang Pan
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
| | - Huaixin Dang
- Zhejiang Yao Yuan Biotechnology Ltd , Jiashan , Zhejiang , China
| | - Xiong Wei
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
| | | | | | - Qiong Zhou
- Biology Department , Wuxi Apptec (Shanghai) Co. Ltd , Shanghai , China
| | | | - Tian Xu
- Shanghai Yao Yuan Biotechnology Ltd (Drug Farm) , Shanghai , China
| |
Collapse
|
9
|
Yoon H, Han J, Jang KL. Hepatitis B Virus X Protein Stimulates Hepatitis C Virus (HCV) Replication by Protecting HCV Core Protein from E6AP-Mediated Proteasomal Degradation. Microbiol Spectr 2022; 10:e0143222. [PMID: 36374094 PMCID: PMC9784765 DOI: 10.1128/spectrum.01432-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Most clinical and experimental studies have suggested that hepatitis C virus (HCV) is dominant over hepatitis B virus (HBV) during coinfection, although the underlying mechanism remains unclear. In this study, we found that the HBV X protein (HBx) upregulates the levels of the HCV core protein to stimulate HCV replication during coinfection in human hepatoma cells. For this purpose, HBx upregulated both the protein levels and enzyme activities of cellular DNA methyltransferase 1 (DNMT1) and DNMT3b, and this subsequently reduced the expression levels of the E6-associated protein (E6AP), an E3 ligase of the HCV core protein, via DNA methylation. The ubiquitin-dependent proteasomal degradation of the HCV core protein was severely impaired in the presence of HBx, whereas this effect was not observed when E6AP was either ectopically expressed or restored by treatment with 5-aza-2'dC or DNMT1 knockdown. The effect of HBx on the HCV core protein was accurately reproduced in HBV/HCV coinfection systems, which were established by either monoinfection by HCV in Huh7D cells transfected with a 1.2-mer HBV replicon or coinfection by HBV and HCV in Huh7D-Na+-taurocholate cotransporting polypeptide cells, providing evidence for the stimulation of HCV replication by HBx. The present study may provide insights into understanding HCV dominance during HBV/HCV coinfection in patients. IMPORTANCE Hepatitis B virus (HBV) and hepatitis C virus (HCV) are major human pathogens that cause a substantial proportion of liver diseases worldwide. As the two hepatotropic viruses have the same modes of transmission, coinfection is often observed, especially in areas and populations where HBV is endemic. High-risk populations include people who inject drugs. Both clinical and experimental studies have shown that HCV is more dominant than HBV during coinfection, but the underlying mechanism remains unclear. In this study, we show that HBV X protein (HBx) stimulates HCV replication by inhibiting the expression of E6-associated protein (E6AP) via DNA methylation, thereby protecting the HCV core protein from proteasomal degradation, which can contribute to HCV dominance during HBV/HCV coinfection.
Collapse
Affiliation(s)
- Hyunyoung Yoon
- Department of Integrated Biological Science, The Graduate School, Pusan National University, Busan, Republic of Korea
| | - Jiwoo Han
- Department of Integrated Biological Science, The Graduate School, Pusan National University, Busan, Republic of Korea
| | - Kyung Lib Jang
- Department of Integrated Biological Science, The Graduate School, Pusan National University, Busan, Republic of Korea
- Department of Microbiology, College of Natural Science, Pusan National University, Busan, Republic of Korea
- Microbiological Resource Research Institute, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
10
|
Skrlec I, Talapko J. Hepatitis B and circadian rhythm of the liver. World J Gastroenterol 2022; 28:3282-3296. [PMID: 36158265 PMCID: PMC9346465 DOI: 10.3748/wjg.v28.i27.3282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/15/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
The circadian rhythm in humans is determined by the central clock located in the hypothalamus’s suprachiasmatic nucleus, and it synchronizes the peripheral clocks in other tissues. Circadian clock genes and clock-controlled genes exist in almost all cell types. They have an essential role in many physiological processes, including lipid metabolism in the liver, regulation of the immune system, and the severity of infections. In addition, circadian rhythm genes can stimulate the immune response of host cells to virus infection. Hepatitis B virus (HBV) infection is the leading cause of liver disease and liver cancer globally. HBV infection depends on the host cell, and hepatocyte circadian rhythm genes are associated with HBV replication, survival, and spread. The core circadian rhythm proteins, REV-ERB and brain and muscle ARNTL-like protein 1, have a crucial role in HBV replication in hepatocytes. In addition to influencing the virus’s life cycle, the circadian rhythm also affects the pharmacokinetics and efficacy of antiviral vaccines. Therefore, it is vital to apply antiviral therapy at the appropriate time of day to reduce toxicity and improve the effectiveness of antiviral treatment. For these reasons, understanding the role of the circadian rhythm in the regulation of HBV infection and host responses to the virus provides us with a new perspective of the interplay of the circadian rhythm and anti-HBV therapy. Therefore, this review emphasizes the importance of the circadian rhythm in HBV infection and the optimization of antiviral treatment based on the circadian rhythm-dependent immune response.
Collapse
Affiliation(s)
- Ivana Skrlec
- Department of Biophysics, Biology, and Chemistry, Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Jasminka Talapko
- Department of Anatomy Histology, Embryology, Pathology Anatomy and Pathology Histology, Faculty of Dental Medicine and Health, Osijek 31000, Croatia
| |
Collapse
|
11
|
Xu M, Lin W, Chung RT. A New Model to Assess Hepatitis B Virus Covalently Closed Circular DNA: A Window Into a Previously Hidden Space? Cell Mol Gastroenterol Hepatol 2022; 13:1255-1256. [PMID: 35085527 PMCID: PMC9073727 DOI: 10.1016/j.jcmgh.2022.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 12/28/2021] [Accepted: 01/03/2022] [Indexed: 12/10/2022]
Affiliation(s)
| | | | - Raymond T. Chung
- Correspondence Address correspondence to: Raymond T. Chung, MD, Liver Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114.
| |
Collapse
|
12
|
Mnyandu N, Limani SW, Arbuthnot P, Maepa MB. Advances in designing Adeno-associated viral vectors for development of anti-HBV gene therapeutics. Virol J 2021; 18:247. [PMID: 34903258 PMCID: PMC8670254 DOI: 10.1186/s12985-021-01715-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/26/2021] [Indexed: 12/25/2022] Open
Abstract
Despite the five decades having passed since discovery of the hepatitis B virus (HBV), together with development of an effective anti-HBV vaccine, infection with the virus remains a serious public health problem and results in nearly 900,000 annual deaths worldwide. Current therapies do not eliminate the virus and viral replication typically reactivates after treatment withdrawal. Hence, current endeavours are aimed at developing novel therapies to achieve a functional cure. Nucleic acid-based therapeutic approaches are promising, with several candidates showing excellent potencies in preclinical and early stages of clinical development. However, this class of therapeutics is yet to become part of standard anti-HBV treatment regimens. Obstacles delaying development of gene-based therapies include lack of clinically relevant delivery methods and a paucity of good animal models for preclinical characterisation. Recent studies have demonstrated safety and efficiency of Adeno-associated viral vectors (AAVs) in gene therapy. However, AAVs do have flaws and this has prompted research aimed at improving design of novel and artificially synthesised AAVs. Main goals are to improve liver transduction efficiencies and avoiding immune clearance. Application of AAVs to model HBV replication in vivo is also useful for characterising anti-HBV gene therapeutics. This review summarises recent advances in AAV engineering and their contributions to progress with anti-HBV gene therapy development.
Collapse
Affiliation(s)
- Njabulo Mnyandu
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shonisani Wendy Limani
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mohube Betty Maepa
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
13
|
Xu Z, Zhao L, Zhong Y, Zhu C, Zhao K, Teng Y, Cheng X, Chen Q, Xia Y. A Novel Mouse Model Harboring Hepatitis B Virus Covalently Closed Circular DNA. Cell Mol Gastroenterol Hepatol 2021; 13:1001-1017. [PMID: 34896285 PMCID: PMC8873614 DOI: 10.1016/j.jcmgh.2021.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS The persistence of viral covalently closed circular DNA (cccDNA) is the major obstacle for antiviral treatment against hepatitis B virus (HBV). Basic and translational studies are largely hampered due to the lack of feasible small animal models to support HBV cccDNA formation. The aim of this study is to establish a novel mouse model harboring cccDNA. METHODS An adeno-associated virus (AAV) vector carrying a replication-deficient HBV1.04-fold genome (AAV-HBV1.04) was constructed. The linear HBV genome starts from nucleotide 403 and ends at 538, which results in the splitting of HBV surface and polymerase genes. Different HBV replication markers were evaluated for AAV-HBV1.04 plasmid-transfected cells, the AAV-HBV1.04 viral vector-transduced cells, and mice injected with the AAV-HBV1.04 viral vector. RESULTS Compared with the previously reported AAV-HBV1.2 construct, direct transfection of AAV-HBV1.04 plasmid failed to produce hepatitis B surface antigen and progeny virus. Interestingly, AAV-HBV1.04 viral vector transduction could result in the formation of cccDNA and the production of all HBV replication markers in vitro and in vivo. The formation of cccDNA could be blocked by ATR (ataxia-telangiectasia and Rad3-related protein) inhibitors but not HBV reverse transcription inhibitor or capsid inhibitors. The AAV-HBV1.04 mouse supported long-term HBV replication and responded to antiviral treatments. CONCLUSIONS This AAV-HBV1.04 mouse model can support HBV cccDNA formation through ATR-mediated DNA damage response. The de novo formed cccDNA but not the parental AAV vector can lead to the production of hepatitis B surface antigen and HBV progeny. This model will provide a unique platform for studying HBV cccDNA and developing novel antivirals against HBV infection.
Collapse
Affiliation(s)
- Zaichao Xu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Li Zhao
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Youquan Zhong
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Chengliang Zhu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kaitao Zhao
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yan Teng
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiaoming Cheng
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China,Wuhan University Center for Pathology and Molecular Diagnostics, Zhongnan Hospital of Wuhan University, Wuhan, China,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Qiang Chen
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yuchen Xia
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China,Correspondence Address correspondence to: Yuchen Xia, PhD, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China. fax: 0086-27-68759222.
| |
Collapse
|
14
|
In Vivo Bioluminescence Imaging of HBV Replicating Hepatocytes Allows for the Monitoring of Anti-Viral Immunity. Viruses 2021; 13:v13112273. [PMID: 34835079 PMCID: PMC8619421 DOI: 10.3390/v13112273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 11/24/2022] Open
Abstract
Immunity against hepatitis B virus (HBV) infection is complex and not entirely understood so far, including the decisive factors leading to the development of chronic hepatitis B. This lack of a mechanistic understanding of HBV-specific immunity is also caused by a limited number of suitable animal models. Here, we describe the generation of a recombinant adenovirus expressing an HBV 1.3-overlength genome linked to luciferase (Ad-HBV-Luc) allowing for precise analysis of the quantity of infected hepatocytes. This enables sensitive and close-meshed monitoring of HBV-specific CD8 T cells and the onset of anti-viral immunity in mice. A high dose of Ad-HBV-Luc developed into chronic hepatitis B accompanied by dysfunctional CD8 T cells characterized by high expression of PD1 and TOX and low expression of KLRG1 and GzmB. In contrast, a low dose of Ad-HBV-Luc infection resulted in acute hepatitis with CD8 T cell-mediated elimination of HBV-replicating hepatocytes associated with elevated sALT levels and increased numbers of cytotoxic HBV-specific CD8 T cells. Thus, the infectious dose was a critical factor to induce either acute self-limited or chronic HBV infection in mice. Taken together, the new Ad-HBV-Luc vector will allow for highly sensitive and time-resolved analysis of HBV-specific immune responses during acute and chronic infection.
Collapse
|
15
|
In Vivo Modelling of Hepatitis B Virus Subgenotype A1 Replication Using Adeno-Associated Viral Vectors. Viruses 2021; 13:v13112247. [PMID: 34835053 PMCID: PMC8618177 DOI: 10.3390/v13112247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/30/2021] [Accepted: 11/04/2021] [Indexed: 12/23/2022] Open
Abstract
The paucity of animal models that simulate the replication of the hepatitis B virus (HBV) is an impediment to advancing new anti-viral treatments. The work reported here employed recombinant adeno-associated viruses (AAVs) to model HBV subgenotype A1 and subgenotype D3 replication in vitro and in vivo. Infection with subgenotype A1 is endemic to parts of sub-Saharan Africa, and it is associated with a high risk of hepatocellular carcinoma. Recombinant AAV serotype 2 (AAV2) and 8 (AAV8) vectors bearing greater-than-genome-length sequences of HBV DNA from subgenotype A1 and D3, were produced. Transduced liver-derived cultured cells produced HBV surface antigen and core antigen. Administration of AAV8 carrying HBV subgenotype A1 genome (AAV8-A1) to mice resulted in the sustained production of HBV replication markers over a six-month period, without elevated inflammatory cytokines, expression of interferon response genes or alanine transaminase activity. Markers of replication were generally higher in animals treated with subgenotype D3 genome-bearing AAVs than in those receiving the subgenotype A1-genome-bearing vectors. To validate the use of the AAV8-A1 murine model for anti-HBV drug development, the efficacy of anti-HBV artificial primary-microRNAs was assessed. Significant silencing of HBV markers was observed over a 6-month period after administering AAVs. These data indicate that AAVs conveniently and safely recapitulate the replication of different HBV subgenotypes, and the vectors may be used to assess antivirals’ potency.
Collapse
|
16
|
Herschke F, Li C, Zhu R, Han Q, Wu Q, Lu Q, Barale-Thomas E, De Jonghe S, Lin TI, De Creus A. JNJ-64794964 (AL-034/TQ-A3334), a TLR7 agonist, induces sustained anti-HBV activity in AAV/HBV mice via non-cytolytic mechanisms. Antiviral Res 2021; 196:105196. [PMID: 34718044 DOI: 10.1016/j.antiviral.2021.105196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/12/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022]
Abstract
JNJ-64794964 (JNJ-4964/AL-034/TQ-A3334), an oral toll-like receptor 7 agonist, is being investigated for the treatment of chronic hepatitis B (CHB), a condition with a high unmet medical need. The anti-hepatitis B (HBV) activity of JNJ-4964 was assessed preclinically in an adeno-associated virus vector expressing HBV (AAV/HBV) mouse model. Mice were treated orally with 2, 6 or 20 mg/kg of JNJ-4964 once-per-week for 12 weeks and then followed up for 4 weeks. At 6 mg/kg, a partial decrease in plasma HBV-DNA and plasma hepatitis B surface antigen (HBsAg) was observed, and anti-HBs antibodies and HBsAg-specific T cells were observed in 1/8 animals. At 20 mg/kg, plasma HBV-DNA and HBsAg levels were undetectable for all animals 3 weeks after start of treatment, with no rebound observed 4 weeks after JNJ-4964 treatment was stopped. High anti-HBs antibody levels were observed until 4 weeks after JNJ-4964 treatment was stopped. In parallel, HBsAg-specific immunoglobulin G-producing B cells and interferon-γ-producing CD4+ T cells were detected in the spleen. In 2/4 animals, liver HBV-DNA and HBV-RNA levels and liver hepatitis B core antigen expression dropped 4 weeks after JNJ-4964 treatment-stop. In these animals, HBsAg-specific CD8+ T cells were detectable. Throughout the study, normal levels of alanine aminotransferase were observed, with no hepatocyte cell death (end of treatment and 4 weeks later) and minimal infiltrations of B and T cells into the liver, suggesting induction of cytokine-mediated, non-cytolytic mechanisms.
Collapse
Affiliation(s)
- Florence Herschke
- Janssen Pharmaceutica NV, 2340, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Chris Li
- Janssen China R&D, Discovery, Shanghai, China
| | - Ren Zhu
- Janssen China R&D, Discovery, Shanghai, China
| | - Qinglin Han
- Janssen China R&D, Discovery, Shanghai, China
| | - Qun Wu
- Janssen China R&D, Discovery, Shanghai, China
| | - Qing Lu
- Janssen China R&D, Discovery, Shanghai, China
| | | | - Sandra De Jonghe
- Janssen Pharmaceutica NV, 2340, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Tse-I Lin
- Janssen Biopharma, 260 E Grand Ave., South San Francisco, CA, 94080, United States
| | - An De Creus
- Janssen Pharmaceutica NV, 2340, Turnhoutseweg 30, 2340, Beerse, Belgium
| |
Collapse
|